1
|
Thangameeran SIM, Tsai ST, Liew HK, Pang CY. Examining Transcriptomic Alterations in Rat Models of Intracerebral Hemorrhage and Severe Intracerebral Hemorrhage. Biomolecules 2024; 14:678. [PMID: 38927081 PMCID: PMC11202056 DOI: 10.3390/biom14060678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening condition associated with significant morbidity and mortality. This study investigates transcriptomic alterations in rodent models of ICH and severe ICH to shed light on the genetic pathways involved in hemorrhagic brain injury. We performed principal component analysis, revealing distinct principal component segments of normal rats compared to ICH and severe ICH rats. We employed heatmaps and volcano plots to identify differentially expressed genes and utilized bar plots and KEGG pathway analysis to elucidate the molecular pathways involved. We identified a multitude of differentially expressed genes in both the ICH and severe ICH models. Our results revealed 5679 common genes among the normal, ICH, and severe ICH groups in the upregulated genes group, and 1196 common genes in the downregulated genes, respectively. A volcano plot comparing these groups further highlighted common genes, including PDPN, TIMP1, SERPINE1, TUBB6, and CD44. These findings underscore the complex interplay of genes involved in inflammation, oxidative stress, and neuronal damage. Furthermore, pathway enrichment analysis uncovered key signaling pathways, including the TNF signaling pathway, protein processing in the endoplasmic reticulum, MAPK signaling pathway, and Fc gamma R-mediated phagocytosis, implicated in the pathogenesis of ICH.
Collapse
Affiliation(s)
| | - Sheng-Tzung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (S.I.M.T.); (S.-T.T.)
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Hock-Kean Liew
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien 97004, Taiwan
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Cheng-Yoong Pang
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (S.I.M.T.); (S.-T.T.)
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| |
Collapse
|
2
|
Koester SW, Hoglund BK, Ciobanu-Caraus O, Hartke JN, Pacult MA, Winkler EA, Catapano JS, Lawton MT. Hematologic and Inflammatory Predictors of Outcome in Patients with Brain Arteriovenous Malformations. World Neurosurg 2024; 185:e342-e350. [PMID: 38340796 DOI: 10.1016/j.wneu.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVE This study investigated the prognostic value of admission blood counts for arteriovenous malformation (AVM) outcomes and compared admission blood counts for patients with ruptured and unruptured AVMs. METHODS A retrospective analysis of patients who underwent surgical treatment for a ruptured cerebral AVM between February 1, 2014, and March 31, 2020, was conducted. The primary outcome was poor neurologic outcome, defined as a modified Rankin Scale score ≥2 in patients with unruptured AVMs or >2 in patients with ruptured AVMs. RESULTS Of 235 included patients, 80 (34%) had ruptured AVMs. At admission, patients with ruptured AVMs had a significantly lower mean (SD) hemoglobin level (12.78 [2.07] g/dL vs. 13.71 [1.60] g/dL, P < 0.001), hematocrit (38.1% [5.9%] vs. 40.7% [4.6%], P < 0.001), lymphocyte count (16% [11%] vs. 26% [10%], P < 0.001), and absolute lymphocyte count (1.41 [0.72] × 103/μL vs. 1.79 [0.68] × 103/μL, P < 0.001), and they had a significantly higher mean (SD) white blood cell count (10.4 [3.8] × 103/μL vs. 7.6 [2.3] × 103/μL, P < 0.001), absolute neutrophil count (7.8 [3.8] × 103/μL vs. 5.0 [2.5] × 103/μL, P < 0.001), and neutrophil count (74% [14%] vs. 64% [13%], P < 0.001). Among patients with unruptured AVMs, white blood cell count ≥6.4 × 103/μL and absolute neutrophil count ≥3.4 × 103/μL were associated with a favorable neurologic outcome, whereas hemoglobin level ≥13.4 g/dL was associated with an unfavorable outcome. Among patients with ruptured AVMs, hypertension was associated with a 3-fold increase in odds of a poor neurologic outcome. CONCLUSIONS Patients with ruptured and unruptured AVMs present with characteristic profiles of hematologic and inflammatory parameters evident in their admission blood work.
Collapse
Affiliation(s)
- Stefan W Koester
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Brandon K Hoglund
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Olga Ciobanu-Caraus
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Joelle N Hartke
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Mark A Pacult
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Ethan A Winkler
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Joshua S Catapano
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Michael T Lawton
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.
| |
Collapse
|
3
|
Wagers ML, Starks A, Nadolski J, Bierbower SM, Altenburg S, Schryer B, Cooper RL. Examining the effect of iron (ferric) on physiological processes: Invertebrate models. Comp Biochem Physiol C Toxicol Pharmacol 2024; 278:109856. [PMID: 38354992 DOI: 10.1016/j.cbpc.2024.109856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/15/2024] [Accepted: 02/04/2024] [Indexed: 02/16/2024]
Abstract
Iron is a common and essential element for maintaining life in bacteria, plants and animals and is found in soil, fresh waters and marine waters; however, over exposure is toxic to organisms. Iron is used in electron transport complexes within mitochondria as well as a co-factor in many essential proteins. It is also established that iron accumulation in the central nervous system in mammals is associated with various neurological disorders. Ample studies have investigated the long-term effects of iron overload in the nervous system. However, its acute effects in nervous tissue and additional organ systems warrant further studies. This study investigates the effects of iron overload on development, behavior, survival, cardiac function, and glutamatergic synaptic transmission in the Drosophila melanogaster. Additionally, physiological responses in crayfish were examined following Fe3+ exposure. Fe3+ reduced neuronal excitability in proprioceptive neurons in a crayfish model. Thus, Fe3+ may block stretch activated channels (SACs) as well as voltage-gated Na+ channels. Exposure also rapidly reduces synaptic transmission but does not block ionotropic glutamatergic receptors, suggesting a blockage of pre-synaptic voltage-gated Ca2+ channels in both crustacean and Drosophila models. The effects are partly reversible with acute exposure, indicating the cells are not rapidly damaged. This study is relevant in demonstrating the effects of Fe3+ on various physiological functions in different organisms in order to further understand the acute and long-term consequences of overload.
Collapse
Affiliation(s)
- Mikaela L Wagers
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA
| | - Ashley Starks
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA
| | - Jeremy Nadolski
- Department of Mathematical and Computational Sciences, Benedictine University, Lisle, IL 60532, USA
| | - Sonya M Bierbower
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Sean Altenburg
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Blake Schryer
- Department of Chemistry and Life Science, United States Military Academy, West Point, NY 10996, USA
| | - Robin L Cooper
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| |
Collapse
|
4
|
Akyol ME, Demir C, Görken G. Investigation of Oxidative Stress Level and Antioxidant Enzyme Activities in Operated and Nonoperated Patients with Spontaneous Intracerebral Hematoma. J Neurol Surg A Cent Eur Neurosurg 2024; 85:21-25. [PMID: 36070790 DOI: 10.1055/a-1938-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
BACKGROUND Complex underlying mechanisms consisting of cytotoxic, excitotoxic, and inflammatory effects of intraparenchymal hemorrhage (ICH) are responsible for the highly detrimental effects on brain tissue. Oxidative stress also plays a significant role in brain damage after ICH; however, it is less important than other factors. In this study, we aimed to evaluate the oxidative stress parameters malondialdehyde (MDA) and antioxidant-reduced glutathione (GSH), superoxide dismutase (SOD), and catalase (CT) activities in operated and nonoperated patients with spontaneous ICH. METHODS One hundred patients with spontaneous ICH and 100 healthy controls were included in this study. Within the indication, 50 of the 100 patients underwent decompressive surgery. MDA, GSH, SOD, and CT activities were measured in the serum obtained from the patients. RESULTS SOD and CT levels were lower in the nonoperated group than in the operated and control groups. GSH was similar in the operated and nonoperated groups, but it was lower in the control group. However, MDA was higher in those who did not undergo surgery than in the other groups. CONCLUSIONS In our study, MDA, an indicator of oxidative stress, was found to be lower, and CT and SOD activities were found to be higher in ICH patients who underwent decompression than in those who did not. This is the first study to present the correlations of MDA, SOD, CT, and GSH in operated and nonoperated patients with spontaneous ICH.
Collapse
Affiliation(s)
- Mehmet Edip Akyol
- Department of Neurosurgery, Van YY University Faculty of Medicine, Van, Turkey
| | - Canan Demir
- Van YY University Vocational School of Health Services, Van, Turkey
| | - Gökhan Görken
- Department of Neurology, Regional Training and Research Hospital, Van, Turkey
| |
Collapse
|
5
|
Paez-Gonzalez P, Lopez-de-San-Sebastian J, Ceron-Funez R, Jimenez AJ, Rodríguez-Perez LM. Therapeutic strategies to recover ependymal barrier after inflammatory damage: relevance for recovering neurogenesis during development. Front Neurosci 2023; 17:1204197. [PMID: 37397456 PMCID: PMC10308384 DOI: 10.3389/fnins.2023.1204197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
The epithelium covering the surfaces of the cerebral ventricular system is known as the ependyma, and is essential for maintaining the physical and functional integrity of the central nervous system. Additionally, the ependyma plays an essential role in neurogenesis, neuroinflammatory modulation and neurodegenerative diseases. Ependyma barrier is severely affected by perinatal hemorrhages and infections that cross the blood brain barrier. The recovery and regeneration of ependyma after damage are key to stabilizing neuroinflammatory and neurodegenerative processes that are critical during early postnatal ages. Unfortunately, there are no effective therapies to regenerate this tissue in human patients. Here, the roles of the ependymal barrier in the context of neurogenesis and homeostasis are reviewed, and future research lines for development of actual therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Patricia Paez-Gonzalez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Raquel Ceron-Funez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
| | - Antonio J. Jimenez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | - Luis Manuel Rodríguez-Perez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
- Department of Human Physiology, Human Histology, Pathological Anatomy and Sports, University of Malaga, Málaga, Spain
| |
Collapse
|
6
|
Wagers ML, Starks A, Abul-Khoudoud MO, Ahmed SM, Alhamdani AW, Ashley C, Bidros PC, Bledsoe CO, Bolton KE, Capili JG, Henning JN, Ison BJ, Moon M, Phe P, Stonecipher SB, Taylor IN, Turner LT, West AK, Cooper RL. An invertebrate model in examining the effect of acute ferric iron exposure on proprioceptive neurons. Comp Biochem Physiol C Toxicol Pharmacol 2023; 266:109558. [PMID: 36717044 DOI: 10.1016/j.cbpc.2023.109558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/11/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Iron is an essential element for plant and animal life and is found in soil, fresh waters and marine waters. The Fe3+ ion is a vital prosthetic group and cofactor to mitochondrial electron transport complexes and numerous proteins involved in normal functioning. Despite its importance to life-sustaining processes, overexposure results in toxicity. For example, ferric iron (Fe3+) accumulation in the mammalian central nervous system is associated with various neurological disorders. Although current literature addresses the long-term effects of Fe3+ overload, fewer studies exist examining the effects of acute exposure. Using the blue crab (Callinectes sapidus), we investigate the effects of acute Fe3+ overload on proprioception within the propodite-dactylopodite (PD) nerve. For proprioceptive studies, 10- and 20-mM ferric chloride and ferric ammonium citrate solutions were used at 5- and 20- min exposure times. Exposure to 20 mM concentrations of ferric chloride and ferric ammonium citrate reduced excitability in proprioceptive neurons. Thus, Fe3+ likely blocks stretch-activated channels or voltage-gated Na+ channels. The depressive effects of Fe3+ are partly reversible following saline washout, indicating cells are not acutely damaged. Gadolinium (GdCl3, 1 and 10 mM) was used to examine the effects of an additional trivalent ion comparator. Gd3+ depressed PD nerve compound action potential amplitude while increasing the compound action potential duration. This study is relevant in demonstrating the dose-dependent effects of acute Fe3+ and Gd3+ exposure on proprioception and provides a model system to further investigate the mechanisms by which metals act on the nervous system.
Collapse
Affiliation(s)
- Mikaela L Wagers
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Ashley Starks
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA
| | | | - Sufia M Ahmed
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | | | - Clair Ashley
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Patrick C Bidros
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | | | - Kayli E Bolton
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Jerone G Capili
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Jamie N Henning
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Bethany J Ison
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Madison Moon
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Panhavuth Phe
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | | | - Isabelle N Taylor
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Logan T Turner
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Aaron K West
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| | - Robin L Cooper
- Department of Biology, University of Kentucky, Lexington 40506, KY, USA.
| |
Collapse
|
7
|
Zheng Y, Li R, Fan X. Targeting Oxidative Stress in Intracerebral Hemorrhage: Prospects of the Natural Products Approach. Antioxidants (Basel) 2022; 11:1811. [PMID: 36139885 PMCID: PMC9495708 DOI: 10.3390/antiox11091811] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH), the second most common subtype of stroke, remains a significant cause of morbidity and mortality worldwide. The pathological mechanism of ICH is very complex, and it has been demonstrated that oxidative stress (OS) plays an important role in the pathogenesis of ICH. Previous studies have shown that OS is a therapeutic target after ICH, and antioxidants have also achieved some benefits in the treatment of ICH. This review aimed to explore the promise of natural products therapy to target OS in ICH. We searched PubMed using the keywords "oxidative stress in intracerebral hemorrhage" and "natural products in intracerebral hemorrhage". Numerous animal and cell studies on ICH have demonstrated the potent antioxidant properties of natural products, including polyphenols and phenolic compounds, terpenoids, alkaloids, etc. In summary, natural products such as antioxidants offer the possibility of treatment of OS after ICH. However, researchers still have a long way to go to apply these natural products for the treatment of ICH more widely in the clinic.
Collapse
Affiliation(s)
| | | | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
8
|
Wang J, Yin J, Zheng X. Artemisinin upregulates neural cell adhesion molecule L1 to attenuate neurological deficits after intracerebral hemorrhage in mice. Brain Behav 2022; 12:e2558. [PMID: 35349764 PMCID: PMC9120716 DOI: 10.1002/brb3.2558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/29/2022] [Accepted: 03/05/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Intracerebral hemorrhage (ICH) is a subtype of stroke and results in neurological deficits in patients without any effective treatments. Artemisinin (ART), a well-known antimalarial Chinese medicine, exerts multiple essential roles in the central and peripheral nervous system due to its antioxidative and anti-inflammation properties. Neural cell adhesion molecule L1 (L1CAM, L1) is considered to be implicated in neural development, functional maintenance, and neuroprotection during disease. However, whether these two essential molecules are neuroprotective in ICH remains unclear. METHODS Therefore, the present study investigated the influence of ART on the recovery of neurological deficits in a mouse model of ICH induced by collagenase and the underlying mechanism. RESULTS It was revealed that ART is capable of upregulating L1 expression to alleviate brain edema, reduce oxidative stress, and inhibit inflammation to alleviate ICH-induced brain injury to improve the neurological outcome in mice suffering from ICH. CONCLUSION These results may lay the foundation for ART to be a novel candidate treatment for ICH.
Collapse
Affiliation(s)
- Jianjiang Wang
- Department of Neurosurgery, General Hospital of Xinjiang Military Region, Urumqi, China
| | - Jie Yin
- Department of Neurosurgery, General Hospital of Xinjiang Military Region, Urumqi, China
| | - Xi Zheng
- Department of Neurosurgery, General Hospital of Xinjiang Military Region, Urumqi, China
| |
Collapse
|
9
|
Magid-Bernstein J, Girard R, Polster S, Srinath A, Romanos S, Awad IA, Sansing LH. Cerebral Hemorrhage: Pathophysiology, Treatment, and Future Directions. Circ Res 2022; 130:1204-1229. [PMID: 35420918 PMCID: PMC10032582 DOI: 10.1161/circresaha.121.319949] [Citation(s) in RCA: 168] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high morbidity and mortality. This review article focuses on the epidemiology, cause, mechanisms of injury, current treatment strategies, and future research directions of ICH. Incidence of hemorrhagic stroke has increased worldwide over the past 40 years, with shifts in the cause over time as hypertension management has improved and anticoagulant use has increased. Preclinical and clinical trials have elucidated the underlying ICH cause and mechanisms of injury from ICH including the complex interaction between edema, inflammation, iron-induced injury, and oxidative stress. Several trials have investigated optimal medical and surgical management of ICH without clear improvement in survival and functional outcomes. Ongoing research into novel approaches for ICH management provide hope for reducing the devastating effect of this disease in the future. Areas of promise in ICH therapy include prognostic biomarkers and primary prevention based on disease pathobiology, ultra-early hemostatic therapy, minimally invasive surgery, and perihematomal protection against inflammatory brain injury.
Collapse
Affiliation(s)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sean Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
10
|
Holste KG, Xia F, Ye F, Keep RF, Xi G. Mechanisms of neuroinflammation in hydrocephalus after intraventricular hemorrhage: a review. Fluids Barriers CNS 2022; 19:28. [PMID: 35365172 PMCID: PMC8973639 DOI: 10.1186/s12987-022-00324-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
Intraventricular hemorrhage (IVH) is a significant cause of morbidity and mortality in both neonatal and adult populations. IVH not only causes immediate damage to surrounding structures by way of mass effect and elevated intracranial pressure; the subsequent inflammation causes additional brain injury and edema. Of those neonates who experience severe IVH, 25-30% will go on to develop post-hemorrhagic hydrocephalus (PHH). PHH places neonates and adults at risk for white matter injury, seizures, and death. Unfortunately, the molecular determinants of PHH are not well understood. Within the past decade an emphasis has been placed on neuroinflammation in IVH and PHH. More information has come to light regarding inflammation-induced fibrosis and cerebrospinal fluid hypersecretion in response to IVH. The aim of this review is to discuss the role of neuroinflammation involving clot-derived neuroinflammatory factors including hemoglobin/iron, peroxiredoxin-2 and thrombin, as well as macrophages/microglia, cytokines and complement in the development of PHH. Understanding the mechanisms of neuroinflammation after IVH may highlight potential novel therapeutic targets for PHH.
Collapse
Affiliation(s)
- Katherine G Holste
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA.
| | - Fan Xia
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 3470 Taubman Center, 1500 E. Medical Center Dr, Ann Arbor, MI, 48109-5338, USA.
- , 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
11
|
Liu J, Zhu Z, Leung GKK. Erythrophagocytosis by Microglia/Macrophage in Intracerebral Hemorrhage: From Mechanisms to Translation. Front Cell Neurosci 2022; 16:818602. [PMID: 35237132 PMCID: PMC8882619 DOI: 10.3389/fncel.2022.818602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition characterized by hematoma related mass effect. Microglia/macrophage (M φ) are rapidly recruited in order to remove the red blood cells through erythrophagocytosis. Efficient erythrophagocytosis can detoxify hemolytic products and facilitate neurological recovery after ICH. The underlying mechanisms include modulation of inflammatory response and oxidative stress, among others. It is a dynamic process mediated by a cascade of signal transduction, including “find-me” signals, “eat-me” signals and a set of phagocytotic receptors-ligand pairs that may be exploited as therapeutic targets. This review summarizes mechanistic signaling pathways of erythrophagocytosis and highlights the potential of harnessing M φ-mediated phagocytosis for ICH treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Zhiyuan Zhu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- Department of Functional Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- *Correspondence: Gilberto Ka-Kit Leung,
| |
Collapse
|
12
|
Zhang Y, Khan S, Liu Y, Zhang R, Li H, Wu G, Tang Z, Xue M, Yong VW. Modes of Brain Cell Death Following Intracerebral Hemorrhage. Front Cell Neurosci 2022; 16:799753. [PMID: 35185473 PMCID: PMC8851202 DOI: 10.3389/fncel.2022.799753] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high rates of mortality and morbidity. It induces cell death that is responsible for neurological deficits postinjury. There are no therapies that effectively mitigate cell death to treat ICH. This review aims to summarize our knowledge of ICH-induced cell death with a focus on apoptosis and necrosis. We also discuss the involvement of ICH in recently described modes of cell death including necroptosis, pyroptosis, ferroptosis, autophagy, and parthanatos. We summarize treatment strategies to mitigate brain injury based on particular cell death pathways after ICH.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhouping Tang
- Department of Neurology, Affiliated Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
- *Correspondence: Mengzhou Xue,
| | - V. Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- V. Wee Yong,
| |
Collapse
|
13
|
Lin J, Tan B, Li Y, Feng H, Chen Y. Sepsis-Exacerbated Brain Dysfunction After Intracerebral Hemorrhage. Front Cell Neurosci 2022; 15:819182. [PMID: 35126060 PMCID: PMC8814659 DOI: 10.3389/fncel.2021.819182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/27/2021] [Indexed: 12/28/2022] Open
Abstract
Sepsis susceptibility is significantly increased in patients with intracerebral hemorrhage (ICH), owing to immunosuppression and intestinal microbiota dysbiosis. To date, ICH with sepsis occurrence is still difficult for clinicians to deal with, and the mortality, as well as long-term cognitive disability, is still increasing. Actually, intracerebral hemorrhage and sepsis are mutually exacerbated via similar pathophysiological mechanisms, mainly consisting of systemic inflammation and circulatory dysfunction. The main consequence of these two processes is neural dysfunction and multiple organ damages, notably, via oxidative stress and neurotoxic mediation under the mediation of central nervous system activation and blood-brain barrier disruption. Besides, the comorbidity-induced multiple organ damages will produce numerous damage-associated molecular patterns and consequently exacerbate the severity of the disease. At present, the prospective views are about operating artificial restriction for the peripheral immune system and achieving cross-tolerance among organs via altering immune cell composition to reduce inflammatory damage.
Collapse
Affiliation(s)
- Jie Lin
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Binbin Tan
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Yuhong Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
| | - Hua Feng
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Hua Feng, ;
| | - Yujie Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing, China
- *Correspondence: Yujie Chen, ;
| |
Collapse
|
14
|
Aronowski J, Sansing LH, Xi G, Zhang JH. Mechanisms of Damage After Cerebral Hemorrhage. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
15
|
Wei Y, Song X, Gao Y, Gao Y, Li Y, Gu L. Iron toxicity in intracerebral hemorrhage: Physiopathological and therapeutic implications. Brain Res Bull 2021; 178:144-154. [PMID: 34838852 DOI: 10.1016/j.brainresbull.2021.11.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023]
Abstract
Intracerebral hemorrhage (ICH)-induced brain injury is a continuous pathological process that involves the deterioration of neurological functions, such as sensory, cognitive or motor functions. Cytotoxic byproducts of red blood cell lysis, especially free iron, appear to be a significant pathophysiologic mechanism leading to ICH-induced injury. Free iron has a crucial role in secondary brain injury after ICH. Chelating iron may attenuate iron-induced neurotoxicity and may be developed as a therapeutic candidate for ICH treatment. In this review, we focused on the potential role of iron toxicity in ICH-induced injury and iron chelation therapy in the management of ICH. It will hopefully advance our understanding of the pathogenesis of ICH and lead to new approaches for treatment.
Collapse
Affiliation(s)
- Yufei Wei
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China
| | - Xiaoxiao Song
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China
| | - Ying Gao
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing 100010, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100010, China
| | - Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100010, China
| | - Lian Gu
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China.
| |
Collapse
|
16
|
Mechanisms of Oxidative Stress and Therapeutic Targets following Intracerebral Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8815441. [PMID: 33688394 PMCID: PMC7920740 DOI: 10.1155/2021/8815441] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/17/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022]
Abstract
Oxidative stress (OS) is induced by the accumulation of reactive oxygen species (ROS) following intracerebral hemorrhage (ICH) and plays an important role in secondary brain injury caused by the inflammatory response, apoptosis, autophagy, and blood-brain barrier (BBB) disruption. This review summarizes the current state of knowledge regarding the pathogenic mechanisms of brain injury after ICH, markers for detecting OS, and therapeutic strategies that target OS to mitigate brain injury.
Collapse
|
17
|
Proteomic Analysis of Perihematoma Tissue from Patients with Intracerebral Hemorrhage Using iTRAQ-Based Quantitative Proteomics. Neuromolecular Med 2021; 23:395-403. [PMID: 33389597 DOI: 10.1007/s12017-020-08637-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/24/2020] [Indexed: 11/27/2022]
Abstract
Intracerebral hemorrhage is a complicated disorder with limited proven prognostic and therapeutic targets and elusive mechanisms. With proteomic methods, we aimed to explore the global protein expression profile of perihematomal tissue from ICH patients and identify potential pathophysiological pathways and protein markers. Using iTRAQ-labeling quantitative proteomics technology, four ICH brain sample and four non-ICH brain samples were analyzed. Among the 3740 quantifiable proteins, 884 were dysregulated in the patients compared to those in the controls (p < 0.05). After bioinformatics analysis, the differentially expressed proteins were found to be mostly involved in hemostatic processes, nutrient metabolism signaling pathways, and antioxidation pathways. Moreover, fibronectin 1 was revealed to be at the center of the protein-protein interaction networks. In summary, the potential pathways and brain protein markers that could potentially be used to predict the prognosis of ICH were obtained from the altered proteomic profile of perihematomal tissue. Thus, these data may yield novel insights into the mechanisms of ICH-induced secondary brain injury.
Collapse
|
18
|
Wang J, Kuang X, Peng Z, Li C, Guo C, Fu X, Wu J, Luo Y, Rao X, Zhou X, Huang B, Tang W, Tang Y. EGCG treats ICH via up-regulating miR-137-3p and inhibiting Parthanatos. Transl Neurosci 2020; 11:371-379. [PMID: 33335777 PMCID: PMC7718614 DOI: 10.1515/tnsci-2020-0143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Intracranial hemorrhage (ICH) causes high mortality and disability without effective treatment in the clinical setting. (-)-Epigallocatechin-3-gallate (EGCG) exerts an essential role in the central nervous system and offers a promising therapeutic agent for the treatment of oxidative damage-related diseases. MiR-137 can inhibit the oxidative stress and apoptosis to attenuate neuronal injury. However, the role of EGCG in regulating miR-137-3p and neuronal Parthanatos remains to be unclear. In the present study, we build the ICH mice model to investigate the antioxidant effects of EGCG via upregulating miR-137-3p and inhibiting neuronal Parthanatos. We revealed that EGCG upregulated miR-137-3p and inhibited neuronal Parthanatos, and promoted the functional recovery, alleviated ICH-induced brain injury, and reduced oxidative stress in mice following ICH. However, following the inhibition of miR-137-3p and activation of Parthanatos, EGCG was unable to exert neuroprotective roles. These combined results suggest that EGCG may upregulate miR-137-3p and inhibit neuronal Parthanatos to accelerate functional recovery in mice after ICH, laying the foundation for EGCG to be a novel strategy for the treatment of neuronal injuries related to Parthanatos.
Collapse
Affiliation(s)
- Jianjun Wang
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
- Department of Clinical, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xuejun Kuang
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Zhao Peng
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Conghui Li
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Chengwu Guo
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xi Fu
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Junhong Wu
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Yang Luo
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xiaolin Rao
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xiangjuan Zhou
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Bin Huang
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Weijun Tang
- Department of Pharmacy, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Yinjuan Tang
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| |
Collapse
|
19
|
Lee EJ, Kee HJ, Han JC, Kee C. Evidence-based understanding of disc hemorrhage in glaucoma. Surv Ophthalmol 2020; 66:412-422. [PMID: 32949554 DOI: 10.1016/j.survophthal.2020.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022]
Abstract
Disc hemorrhage is a characteristic finding that is highly associated with glaucoma development or progression. Consequently, the literature commonly designates disc hemorrhage as a "risk factor" for glaucoma progression; however, the exact cause-and-effect relationship or mechanism remains unclear. In this review, we discuss the emerging evidence that disc hemorrhage is a secondary development that follows glaucomatous damage. As our understanding of disc hemorrhage has progressed in recent decades, we suggest the terminology be changed from "risk factor" to "indicator" of ongoing glaucomatous development or progression for a more accurate description, better indication of the clinical implications and, ultimately, a better guide for future research.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Joo Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong Chul Han
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Changwon Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Tada S, Shiota A, Hayashi H, Nakamura T. Reference urinary biopyrrin level and physiological variation in healthy young adults: relation of stress by learning. Heliyon 2020; 6:e03138. [PMID: 32042943 PMCID: PMC7002780 DOI: 10.1016/j.heliyon.2019.e03138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/22/2019] [Accepted: 12/27/2019] [Indexed: 11/25/2022] Open
Abstract
Background Biopyrrins are end products of oxidation reactions of bilirubin with reactive oxygen, and urinary biopyrrin (UBP) levels might increase under oxidative stress. The authors examined the reference UBP level for healthy adults and its physiological variation in 40 healthy volunteers recruited from among students of our university (20 students each from third-year and fourth-year), and compared the results with data on 8-hydroxy-2-deoxyguanosine (8OHdG). Methods UBP and 8OHdG levels could be considered as oxidative stress markers. The UBP levels were measured with a competitive ELISA kit using biopyrrin antibody 24G7, according to the manufacturer's protocol. 8OHdG levels were measured with a Highly Sensitive 8-OHdG Check kit. UBP and 8OHdG measurements were performed in triplicate and means values calculated. For both parameters, creatinine (Cr) correction was performed using urinary creatinine levels measured by an enzymatic method. Results A comparison of the UBP levels between different grades revealed that the third-year students under high stress from clinical training and other course work tended to have a higher UBP level than fourth-year students. Therefore, we compared the current UBP levels in fourth-year students (samples collected in 2018) with their UBP level when they were in the third-year (samples collected in 2017) to examine the annual change. We found that the UBP level in 2017 samples was significantly higher than that in 2018 samples (P < 0.05). No difference in the 8OHdG level. Additionally, no effect of menstrual stress on the UBP level was observed. Conclusions These results suggest that the UBP levels may be related to school-related stress and menstruation has no effects on urinalysis results.
Collapse
Affiliation(s)
- Satoshi Tada
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1 Hara Mure-cho, Takamatsu City, Kagawa 761-0123, Japan
| | - Atsuko Shiota
- Department of Nursing, Kagawa Prefectural University of Health Sciences, 281-1 Hara Mure-cho, Takamatsu City, Kagawa 761-0123, Japan
| | - Hidehiro Hayashi
- Clinical Department of Research and Development, Cellspect Co. Ltd., Morioka, 020-0857, Japan
| | - Takehiro Nakamura
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1 Hara Mure-cho, Takamatsu City, Kagawa 761-0123, Japan
| |
Collapse
|
21
|
Gastrodin Attenuates Neuronal Apoptosis and Neurological Deficits after Experimental Intracerebral Hemorrhage. J Stroke Cerebrovasc Dis 2019; 29:104483. [PMID: 31727597 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104483] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/24/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Gastrodin, a glucoside of gastrodigenin, inhibits cerebral oxidant stress and apoptosis in multiple central nervous system injury, but its effect in intracerebral hemorrhage (ICH) remains unclear. This study investigated the effect of gastrodin on neuronal apoptosis and neurological deficits in rat ICH model. METHODS In vitro experiments were performed using hematoma lysate-induced cell damage model in primary cortical neurons. Rat ICH model was produced by a caudatum injection of collagenase. Gastrodin was intraperitoneal injected after 2 hours following ICH. Cell viability, brain water content, neurological score, western blot, and immunofluorescence experiments were performed. RESULTS Gastrodin significantly decreased hematoma lysate-induced reduction of cell viability and cell apoptosis in primary cortical neurons. Gastrodin significantly improved brain edema and neurological deficits post-ICH. Moreover, gastrodin administration significantly reduced levels of ROS, 8-OHDG, 3-Nitrotyrosine and MDA, while increased GSH-Px and SOD activity, and stimulated the upregulation of Keap1, Nrf2, and HO-1 signaling at 72 hours post-ICH. Furthermore, gastrodin significantly increased Bcl-2 expression, while reduced level of Bax, active caspase-3 and active caspase-9, also reduced the number of active caspase-3 or TUNEL positive neurons at 72 hours post-ICH. CONCLUSION These results suggest that gastrodin is neuroprotective after ICH and the mechanism may be associated with the inhibition of oxidative stress and neuronal apoptosis.
Collapse
|
22
|
Neuroprotection for ischemic stroke in the endovascular era: A brief report on the future of intra-arterial therapy. J Clin Neurosci 2019; 69:289-291. [DOI: 10.1016/j.jocn.2019.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/05/2019] [Indexed: 01/05/2023]
|
23
|
Yauger YJ, Bermudez S, Moritz KE, Glaser E, Stoica B, Byrnes KR. Iron accentuated reactive oxygen species release by NADPH oxidase in activated microglia contributes to oxidative stress in vitro. J Neuroinflammation 2019; 16:41. [PMID: 30777083 PMCID: PMC6378754 DOI: 10.1186/s12974-019-1430-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/04/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Excessive iron contributes to oxidative stress after central nervous system injury. NADPH oxidase (NOX) enzymes are upregulated in microglia after pro-inflammatory activation and contribute to oxidative stress. The relationship between iron, microglia, NOX, and oxidative stress is currently unclear. METHODS We evaluated the effects of iron on lipopolysaccharide (LPS)-activated microglia and its secondary effect within neuronal co-cultures. Further, NOX2 and four specific inhibitors were tested to evaluate the relationship with the reactive oxygen species (ROS)-producing enzymes. RESULTS An iron dose-dependent increase in ROS production among microglia treated with LPS was identified. Interestingly, despite this increase in ROS, inflammatory polarization alterations were not detected among the microglia after exposure to iron and LPS. Co-culture experimentation between primary neurons and exposed microglia (iron and LPS) significantly reduced neuronal cell number at 24 h, suggesting a profound neurotoxic effect despite the lack of a change in polarization phenotype. NOX2 and NOX4 inhibition significantly reduced ROS production among microglia exposed to iron and LPS and reduced neuronal damage and death in response to microglial co-culture. CONCLUSIONS In conclusion, iron significantly increased ROS production and neurotoxicity without exacerbating LP-activated microglia phenotype in vitro, suggesting that iron contributes to microglia-related oxidative stress, and this may be a viable therapeutic target for injury or neurodegeneration. Further, this study highlights both NOX2 and NOX4 as potential therapeutic targets in the treatment of iron-induced microglia-related inflammation and neurotoxicity.
Collapse
Affiliation(s)
- Young J Yauger
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Sara Bermudez
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kasey E Moritz
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Ethan Glaser
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland, School of Medicine, 655 W. Baltimore St, Room #6-015, Baltimore, MD, USA
| | - Bogdan Stoica
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland, School of Medicine, 655 W. Baltimore St, Room #6-015, Baltimore, MD, USA
| | - Kimberly R Byrnes
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Room C2099, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
24
|
Vaibhav K, Braun M, Khan MB, Fatima S, Saad N, Shankar A, Khan ZT, Harris RBS, Yang Q, Huo Y, Arbab AS, Giri S, Alleyne CH, Vender JR, Hess DC, Baban B, Hoda MN, Dhandapani KM. Remote ischemic post-conditioning promotes hematoma resolution via AMPK-dependent immune regulation. J Exp Med 2018; 215:2636-2654. [PMID: 30190288 PMCID: PMC6170180 DOI: 10.1084/jem.20171905] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 05/07/2018] [Accepted: 08/17/2018] [Indexed: 01/16/2023] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) produces the highest acute mortality and worst outcomes of all stroke subtypes. Hematoma volume is an independent determinant of ICH patient outcomes, making clot resolution a primary goal of clinical management. Herein, remote-limb ischemic post-conditioning (RIC), the repetitive inflation-deflation of a blood pressure cuff on a limb, accelerated hematoma resolution and improved neurological outcomes after ICH in mice. Parabiosis studies revealed RIC accelerated clot resolution via a humoral-mediated mechanism. Whereas RIC increased anti-inflammatory macrophage activation, myeloid cell depletion eliminated the beneficial effects of RIC after ICH. Myeloid-specific inactivation of the metabolic regulator, AMPKα1, attenuated RIC-induced anti-inflammatory macrophage polarization and delayed hematoma resolution, providing a molecular link between RIC and immune activation. Finally, chimera studies implicated myeloid CD36 expression in RIC-mediated neurological recovery after ICH. Thus, RIC, a clinically well-tolerated therapy, noninvasively modulates innate immune responses to improve ICH outcomes. Moreover, immunometabolic changes may provide pharmacodynamic blood biomarkers to clinically monitor the therapeutic efficacy of RIC.
Collapse
Affiliation(s)
- Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | | | - Sumbul Fatima
- Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA
| | - Nancy Saad
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA
| | - Adarsh Shankar
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Zenab T Khan
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Ruth B S Harris
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Qiuhua Yang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA
| | - Yuqing Huo
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA
| | - Ali S Arbab
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI
| | - Cargill H Alleyne
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Babak Baban
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Md Nasrul Hoda
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA
- Department of Medical Laboratory, Imaging, and Radiological Sciences, College of Allied Health Sciences, Augusta University, Augusta, GA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
25
|
Qiao HB, Li J, Lv LJ, Nie BJ, Lu P, Xue F, Zhang ZM. Eupatilin inhibits microglia activation and attenuates brain injury in intracerebral hemorrhage. Exp Ther Med 2018; 16:4005-4009. [PMID: 30344678 PMCID: PMC6176204 DOI: 10.3892/etm.2018.6699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 05/17/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammation serves a critical role in the pathophysiology of intracerebral hemorrhage (ICH)-induced brain injury. Eupatilin, a pharmacologically active flavone derived from Artemisia sp., has been reported to have antioxidant, anti-inflammatory, anti-allergic and antitumor activities. However, the effect of eupatilin in ICH has not been well studied. The aim of the present study was to investigate the effect of eupatilin on ICH-induced microglial inflammation. The MTT and Transwell migration assay results revealed that eupatilin significantly inhibited microglial migration. It also decreased the production of inflammatory cytokines in erythrocyte lysis-induced BV2 cells, as well as the level of intracellular reactive oxygen species. The anti-inflammatory mechanism of eupatilin was also investigated using ELISAs and western blotting and the results demonstrated that eupatilin was able to inhibit erythrocyte lysis-induced NF-κB activation in BV2 cells. Taken together, the results of the present study suggest that eupatilin serves neurological protective effects via inhibiting microglial inflammation, providing an experimental basis for the use of eupatilin as a therapeutic target for ICH.
Collapse
Affiliation(s)
- Hai-Bo Qiao
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Jia Li
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Lian-Jie Lv
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Ben-Jin Nie
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Peng Lu
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Feng Xue
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| | - Zhi-Ming Zhang
- Department of Neurosurgery, Tianjin Nankai Hospital, Tianjin 300100, P.R. China
| |
Collapse
|
26
|
Zhao X, Ting SM, Sun G, Roy-O'Reilly M, Mobley AS, Bautista Garrido J, Zheng X, Obertas L, Jung JE, Kruzel M, Aronowski J. Beneficial Role of Neutrophils Through Function of Lactoferrin After Intracerebral Hemorrhage. Stroke 2018; 49:1241-1247. [PMID: 29636422 DOI: 10.1161/strokeaha.117.020544] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/13/2018] [Accepted: 02/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND PURPOSE Intracerebral hemorrhage (ICH) is a devastating disease with a 30-day mortality of ~50%. There are no effective therapies for ICH. ICH results in brain damage in 2 major ways: through the mechanical forces of extravasated blood and then through toxicity of the intraparenchymal blood components including hemoglobin/iron. LTF (lactoferrin) is an iron-binding protein, uniquely abundant in polymorphonuclear neutrophils (PMNs). After ICH, circulating blood PMNs enter the ICH-afflicted brain where they release LTF. By virtue of sequestrating iron, LTF may contribute to hematoma detoxification. METHODS ICH in mice was produced using intrastriatal autologous blood injection. PMNs were depleted with intraperitoneal administration of anti-Ly-6G antibody. Treatment of mouse brain cell cultures with lysed RBC or iron was used as in vitro model of ICH. RESULTS LTF mRNA was undetectable in the mouse brain, even after ICH. Unlike mRNA, LTF protein increased in ICH-affected hemispheres by 6 hours, peaked at 24 to 72 hours, and remained elevated for at least a week after ICH. At the single cell level, LTF was detected in PMNs in the hematoma-affected brain at all time points after ICH. We also found elevated LTF in the plasma after ICH, with a temporal profile similar to LTF changes in the brain. Importantly, mrLTF (recombinant mouse LTF) reduced the cytotoxicity of lysed RBC and FeCl3 to brain cells in culture. Ultimately, in an ICH model, systemic administration of mrLTF (at 3, 24, and 48 hours after ICH) reduced brain edema and ameliorated neurological deficits caused by ICH. mrLTF retained the benefit in reducing behavioral deficit even with 24-hour treatment delay. Interestingly, systemic depletion of PMNs at 24 hours after ICH worsened neurological deficits, suggesting that PMN infiltration into the brain at later stages after ICH could be a beneficial response. CONCLUSIONS LTF delivered to the ICH-affected brain by infiltrating PMNs may assist in hematoma detoxification and represent a powerful potential target for the treatment of ICH.
Collapse
Affiliation(s)
- Xiurong Zhao
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Shun-Ming Ting
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Guanghua Sun
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Meaghan Roy-O'Reilly
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Alexis S Mobley
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Jesus Bautista Garrido
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Xueping Zheng
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Lidiya Obertas
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Joo Eun Jung
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| | - Marian Kruzel
- Department of Integrative Biology and Pharmacology (M.K.), McGovern Medical School, University of Texas HSC, Houston
| | - Jaroslaw Aronowski
- From the Department of Neurology (X.Z., S.-M.T., G.S., M.R.-O., A.S.M., J.B.G., X.Z., L.O., J.E.J., J.A.)
| |
Collapse
|
27
|
Haque ME, Gabr RE, Zhao X, Hasan KM, Valenzuela A, Narayana PA, Ting SM, Sun G, Savitz SI, Aronowski J. Serial quantitative neuroimaging of iron in the intracerebral hemorrhage pig model. J Cereb Blood Flow Metab 2018; 38:375-381. [PMID: 29292651 PMCID: PMC5851147 DOI: 10.1177/0271678x17751548] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Iron released after intracerebral hemorrhage (ICH) is damaging to the brain. Measurement of the content and distribution of iron in the hematoma could predict brain damage. In this study, 16 Yorkshire piglets were subjected to autologous blood injection ICH model and studied longitudinally using quantitative susceptibility mapping and R2* relaxivity MRI on day 1 and 7 post-ICH. Phantom calibration of susceptibility demonstrated (1) iron distribution heterogeneity within the hematoma and (2) natural absorption of iron from 154 ± 78 µg/mL (day 1) to 127 ± 33 µg/mL (day 7). R2* in the hematoma decreased at day 7. This method could be adopted for ICH in humans.
Collapse
Affiliation(s)
- Muhammad E Haque
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Refaat E Gabr
- 2 Diagnostic and Interventional Imaging at UTHealth, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiurong Zhao
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Khader M Hasan
- 2 Diagnostic and Interventional Imaging at UTHealth, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andrew Valenzuela
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ponnada A Narayana
- 2 Diagnostic and Interventional Imaging at UTHealth, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shun-Ming Ting
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Guanghua Sun
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sean I Savitz
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jaroslaw Aronowski
- 1 Institute for Stroke and Cerebrovascular Disease, McGovern 12339 Medical School at UTHealth , University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
28
|
Melatonin: A Versatile Protector against Oxidative DNA Damage. Molecules 2018; 23:molecules23030530. [PMID: 29495460 PMCID: PMC6017920 DOI: 10.3390/molecules23030530] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/13/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022] Open
Abstract
Oxidative damage to DNA has important implications for human health and has been identified as a key factor in the onset and development of numerous diseases. Thus, it is evident that preventing DNA from oxidative damage is crucial for humans and for any living organism. Melatonin is an astonishingly versatile molecule in this context. It can offer both direct and indirect protection against a wide variety of damaging agents and through multiple pathways, which may (or may not) take place simultaneously. They include direct antioxidative protection, which is mediated by melatonin's free radical scavenging activity, and also indirect ways of action. The latter include, at least: (i) inhibition of metal-induced DNA damage; (ii) protection against non-radical triggers of oxidative DNA damage; (iii) continuous protection after being metabolized; (iv) activation of antioxidative enzymes; (v) inhibition of pro-oxidative enzymes; and (vi) boosting of the DNA repair machinery. The rather unique capability of melatonin to exhibit multiple neutralizing actions against diverse threatening factors, together with its low toxicity and its ability to cross biological barriers, are all significant to its efficiency for preventing oxidative damage to DNA.
Collapse
|
29
|
Grunwald Z, Beslow LA, Urday S, Vashkevich A, Ayres A, Greenberg SM, Goldstein JN, Leasure A, Shi FD, Kahle KT, Battey TWK, Simard JM, Rosand J, Kimberly WT, Sheth KN. Perihematomal Edema Expansion Rates and Patient Outcomes in Deep and Lobar Intracerebral Hemorrhage. Neurocrit Care 2017; 26:205-212. [PMID: 27844466 DOI: 10.1007/s12028-016-0321-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Perihematomal edema (PHE) expansion rate may predict functional outcome following spontaneous intracerebral hemorrhage (ICH). We hypothesized that the effect of PHE expansion rate on outcome is greater for deep versus lobar ICH. METHODS Subjects (n = 115) were retrospectively identified from a prospective ICH cohort enrolled from 2000 to 2013. Inclusion criteria were age ≥ 18 years, spontaneous supratentorial ICH, and known onset time. Exclusion criteria were primary intraventricular hemorrhage (IVH), trauma, subsequent surgery, or warfarin-related ICH. ICH and PHE volumes were measured from CT scans and used to calculate expansion rates. Logistic regression assessed the association between PHE expansion rates and 90-day mortality or poor functional outcome (modified Rankin Scale > 2). Odds ratios are per 0.04 mL/h. RESULTS PHE expansion rate from baseline to 24 h (PHE24) was associated with mortality for deep (p = 0.03, OR 1.13[1.02-1.26]) and lobar ICH (p = 0.02, OR 1.03[1.00-1.06]) in unadjusted regression and in models adjusted for age (deep p = 0.02, OR 1.15[1.02-1.28]; lobar p = 0.03, OR 1.03[1.00-1.06]), Glasgow Coma Scale (deep p = 0.03, OR 1.13[1.01-1.27]; lobar p = 0.02, OR 1.03[1.01-1.06]), or time to baseline CT (deep p = 0.046, OR 1.12[1.00-1.25]; lobar p = 0.047, OR 1.03[1.00-1.06]). PHE expansion rate from baseline to 72 h (PHE72) was associated with mRS > 2 for deep ICH in models that were unadjusted (p = 0.02, OR 4.04[1.25-13.04]) or adjusted for ICH volume (p = 0.02, OR 4.3[1.25-14.98]), age (p = 0.03, OR 5.4[1.21-24.11]), GCS (p = 0.02, OR 4.19[1.2-14.55]), or time to first CT (p = 0.03, OR 4.02[1.19-13.56]). CONCLUSIONS PHE72 was associated with poor functional outcomes after deep ICH, whereas PHE24 was associated with mortality for deep and lobar ICH.
Collapse
Affiliation(s)
- Zachary Grunwald
- Department of Neurology, Yale School of Medicine, 15 York Street, Bldg. LLCI, 10th Floor, 1003C, New Haven, CT, 06510, USA.
| | - Lauren A Beslow
- Department of Neurology, Yale School of Medicine, 15 York Street, Bldg. LLCI, 10th Floor, 1003C, New Haven, CT, 06510, USA
| | - Sebastian Urday
- Department of Neurology, Yale School of Medicine, 15 York Street, Bldg. LLCI, 10th Floor, 1003C, New Haven, CT, 06510, USA
| | - Anastasia Vashkevich
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alison Ayres
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua N Goldstein
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Audrey Leasure
- Department of Neurology, Yale School of Medicine, 15 York Street, Bldg. LLCI, 10th Floor, 1003C, New Haven, CT, 06510, USA
| | - Fu-Dong Shi
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Thomas W K Battey
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Marc Simard
- Departments of Neurosurgery, Pathology and Physiology, University of Maryland School of Medicine, Baltimore, MA, USA
| | - Jonathan Rosand
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - W Taylor Kimberly
- Center for Human Genetic Research and Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kevin N Sheth
- Department of Neurology, Yale School of Medicine, 15 York Street, Bldg. LLCI, 10th Floor, 1003C, New Haven, CT, 06510, USA
| |
Collapse
|
30
|
Wilkinson DA, Pandey AS, Thompson BG, Keep RF, Hua Y, Xi G. Injury mechanisms in acute intracerebral hemorrhage. Neuropharmacology 2017; 134:240-248. [PMID: 28947377 DOI: 10.1016/j.neuropharm.2017.09.033] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/21/2017] [Indexed: 10/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is the most common hemorrhagic stroke subtype, and rates are increasing with an aging population. Despite an increase in research and trials of therapies for ICH, mortality remains high and no interventional therapy has been demonstrated to improve outcomes. We review known mechanisms of injury, recent clinical trial results, and newly discovered signaling pathways involved in hematoma clearance. Enthusiasm remains high for methods of minimally invasive clot removal as well as pharmacologic strategies to improve recovery after ICH, both of which are currently being evaluated in clinical trials. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
| | - Aditya S Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
31
|
Silymarin prevents NLRP3 inflammasome activation and protects against intracerebral hemorrhage. Biomed Pharmacother 2017. [DOI: 10.1016/j.biopha.2017.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
32
|
Wang Z, Zhou F, Dou Y, Tian X, Liu C, Li H, Shen H, Chen G. Melatonin Alleviates Intracerebral Hemorrhage-Induced Secondary Brain Injury in Rats via Suppressing Apoptosis, Inflammation, Oxidative Stress, DNA Damage, and Mitochondria Injury. Transl Stroke Res 2017; 9:74-91. [PMID: 28766251 PMCID: PMC5750335 DOI: 10.1007/s12975-017-0559-x] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/20/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022]
Abstract
Intracerebral hemorrhage (ICH) is a cerebrovascular disease with high mortality and morbidity, and the effective treatment is still lacking. We designed this study to investigate the therapeutic effects and mechanisms of melatonin on the secondary brain injury (SBI) after ICH. An in vivo ICH model was induced via autologous whole blood injection into the right basal ganglia in Sprague-Dawley (SD) rats. Primary rat cortical neurons were treated with oxygen hemoglobin (OxyHb) as an in vitro ICH model. The results of the in vivo study showed that melatonin alleviated severe brain edema and behavior disorders induced by ICH. Indicators of blood-brain barrier (BBB) integrity, DNA damage, inflammation, oxidative stress, apoptosis, and mitochondria damage showed a significant increase after ICH, while melatonin reduced their levels. Meanwhile, melatonin promoted further increasing of expression levels of antioxidant indicators induced by ICH. Microscopically, TUNEL and Nissl staining showed that melatonin reduced the numbers of ICH-induced apoptotic cells. Inflammation and DNA damage indicators exhibited an identical pattern compared to those above. Additionally, the in vitro study demonstrated that melatonin reduced the apoptotic neurons induced by OxyHb and protected the mitochondrial membrane potential. Collectively, our investigation showed that melatonin ameliorated ICH-induced SBI by impacting apoptosis, inflammation, oxidative stress, DNA damage, brain edema, and BBB damage and reducing mitochondrial membrane permeability transition pore opening, and melatonin may be a potential therapeutic agent of ICH.
Collapse
Affiliation(s)
- Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Feng Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Yang Dou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Xiaodi Tian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Chenglin Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
| |
Collapse
|
33
|
Xiong XY, Liu L, Wang FX, Yang YR, Hao JW, Wang PF, Zhong Q, Zhou K, Xiong A, Zhu WY, Zhao T, Meng ZY, Wang YC, Gong QW, Liao MF, Wang J, Yang QW. Toll-Like Receptor 4/MyD88-Mediated Signaling of Hepcidin Expression Causing Brain Iron Accumulation, Oxidative Injury, and Cognitive Impairment After Intracerebral Hemorrhage. Circulation 2016; 134:1025-1038. [PMID: 27576776 DOI: 10.1161/circulationaha.116.021881] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Disturbance of brain iron metabolism after intracerebral hemorrhage (ICH) results in oxidative brain injury and cognition impairment. Hepcidin plays an important role in regulating iron metabolism, and we have reported that serum hepcidin is positively correlated with poor outcomes in patients with ICH. However, the roles of hepcidin in brain iron metabolism after ICH remain largely unknown. METHODS Parabiosis and ICH models combined with in vivo and in vitro experiments were used to investigate the roles of hepcidin in brain iron metabolism after ICH. RESULTS Increased hepcidin-25 was found in serum and primarily in astrocytes after ICH. The brain iron efflux, oxidative brain injury, and cognition impairment were improved in Hepc-/- ICH mice but aggravated by the human hepcidin-25 peptide in C57BL/6 ICH mice. Data obtained in in vitro studies showed that increased hepcidin inhibited the intracellular iron efflux of brain microvascular endothelial cells but was rescued by a hepcidin antagonist, fursultiamine. Using parabiosis ICH models also shows that increased serum hepcidin prevents brain iron efflux. In addition, Toll-like receptor 4 (TLR4)/MyD88 signaling pathway increased hepcidin expression by promoting interleukin-6 expression and signal transducer and activator of transcription 3 phosphorylation. TLR4-/- and MyD88-/- mice exhibited improvement in brain iron efflux at 7, 14, and 28 days after ICH, and the TLR4 antagonist (6R)-6-[N-(2-chloro-4-fluorophenyl) sulfamoyl] cyclohex-1-ene-1-carboxylate significantly decreased brain iron levels at days 14 and 28 after ICH and improved cognition impairment at day 28. CONCLUSIONS The results presented here show that increased hepcidin expression caused by inflammation prevents brain iron efflux via inhibition of the intracellular iron efflux of brain microvascular endothelial cells entering into circulation and aggravating oxidative brain injury and cognition impairment, which identifies a mechanistic target for muting inflammation to promote brain iron efflux and to attenuate oxidative brain injury after ICH.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Liang Liu
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Fa-Xiang Wang
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Yuan-Rui Yang
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Jun-Wei Hao
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Peng-Fei Wang
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Qi Zhong
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Kai Zhou
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Ao Xiong
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Wen-Yao Zhu
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Ting Zhao
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Zhao-You Meng
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Yan-Chun Wang
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Qiu-Wen Gong
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Mao-Fan Liao
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Jian Wang
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.)
| | - Qing-Wu Yang
- From Department of Neurology, Xinqiao Hospital, Third Military Medical University, Shapingba District, Chongqing, China (X.-Y.X., L.L., F.-X.W., Y.-R.Y., Q.Z., K.Z., W.-Y.Z., T.Z., Z.-Y.M., Y.-C.W., Q.-W.G., M.-F.L., Q.-W.Y.); Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China (J.-W.H.); Department of Neurology, Weihai Municipal Hospital, Weihai, China (P.-F.W.); Basic Medical College, Zhengzhou University, Zhengzhou, China (A.X.); and Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD (J.W.).
| |
Collapse
|
34
|
Chen M, Lai L, Li X, Zhang X, He X, Liu W, Li R, Ke X, Fu C, Huang Z, Duan C. Baicalein Attenuates Neurological Deficits and Preserves Blood-Brain Barrier Integrity in a Rat Model of Intracerebral Hemorrhage. Neurochem Res 2016; 41:3095-3102. [PMID: 27518088 DOI: 10.1007/s11064-016-2032-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/12/2016] [Accepted: 08/05/2016] [Indexed: 01/19/2023]
Abstract
Previous studies have demonstrated that baicalein has protective effects against several diseases, which including ischemic stroke. The effect of baicalein on the blood-brain barrier (BBB) in intracerebral hemorrhage (ICH) and its related mechanisms are not well understood. We aimed to investigate the mechanisms by which baicalein may influence the BBB in a rat model of ICH. The rat model of ICH was induced by intravenous injection of collagenase IV into the brain. Animals were randomly divided into three groups: sham operation, vehicle, and baicalein group. Each group was then divided into subgroups, in which the rats were sacrificed at 24 and 72 h after ICH. We assessed brain edema, behavioral changes, BBB leakage, apoptosis, inducible nitric oxide synthase (iNOS), zonula occludens (ZO)-1, Mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB). Treatment with baicalein reduced brain water content, BBB leakage, apoptosis, and neurologic deficits, compared with vehicle. Baicalein also decreased ICH-induced changes in the levels of iNOS but increased the levels of ZO-1. The protective effect of baicalein on the BBB in ICH rats was possibly invoked by attenuated p-38 MAPK and JNK phosphorylation, and decreased activation of the NF-κB signaling pathway, which may have suppressed gene transcription, including iNOS, and eventually decreased formation of peroxynitrite (ONOO-). Our results suggest that baicalein exerts a protective effect on BBB disruption in the rat model of ICH. The likely mechanism is via inhibition of MAPKs and NF-κB signaling pathways, leading to decreased formation of iNOS and ONOO-, thereby improving neurological function.
Collapse
Affiliation(s)
- Min Chen
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Lingfeng Lai
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xifeng Li
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xin Zhang
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xuying He
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Wenchao Liu
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Ran Li
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xunchang Ke
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Chuanyi Fu
- Department of Neurosurgery, People's Hospital of Hainan Province, Haikou, Hainan, China
| | - Zhiwei Huang
- Department of Neurosurgery, Liuzhou Worker's Hospital, Guangxi, China
| | - Chuanzhi Duan
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
35
|
Glushakov AV, Arias RA, Tolosano E, Doré S. Age-Dependent Effects of Haptoglobin Deletion in Neurobehavioral and Anatomical Outcomes Following Traumatic Brain Injury. Front Mol Biosci 2016; 3:34. [PMID: 27486583 PMCID: PMC4949397 DOI: 10.3389/fmolb.2016.00034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/05/2016] [Indexed: 12/11/2022] Open
Abstract
Cerebral hemorrhages are common features of traumatic brain injury (TBI) and their presence is associated with chronic disabilities. Recent clinical and experimental evidence suggests that haptoglobin (Hp), an endogenous hemoglobin-binding protein most abundant in blood plasma, is involved in the intrinsic molecular defensive mechanism, though its role in TBI is poorly understood. The aim of this study was to investigate the effects of Hp deletion on the anatomical and behavioral outcomes in the controlled cortical impact model using wildtype (WT) C57BL/6 mice and genetically modified mice lacking the Hp gene (Hp(-∕-)) in two age cohorts [2-4 mo-old (young adult) and 7-8 mo-old (older adult)]. The data obtained suggest age-dependent significant effects on behavioral and anatomical TBI outcomes and recovery from injury. Moreover, in the adult cohort, neurological deficits in Hp(-∕-) mice at 24 h were significantly improved compared to WT, whereas there were no significant differences in brain pathology between these genotypes. In contrast, in the older adult cohort, Hp(-∕-) mice had significantly larger lesion volumes compared to WT, but neurological deficits were not significantly different. Immunohistochemistry for ionized calcium-binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein (GFAP) revealed significant differences in microglial and astrocytic reactivity between Hp(-∕-) and WT in selected brain regions of the adult but not the older adult-aged cohort. In conclusion, the data obtained in the study provide clarification on the age-dependent aspects of the intrinsic defensive mechanisms involving Hp that might be involved in complex pathways differentially affecting acute brain trauma outcomes.
Collapse
Affiliation(s)
- Alexander V Glushakov
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| | - Rodrigo A Arias
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| | - Emanuela Tolosano
- Departments of Molecular Biotechnology and Health Sciences, University of Torino Torino, Italy
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of MedicineGainesville, FL, USA; Departments of Anesthesiology, Neurology, Psychiatry, Psychology, Pharmaceutics and Neuroscience, University of Florida College of MedicineGainesville, FL, USA
| |
Collapse
|
36
|
Obermann M, Gizewski ER, Limmroth V, Diener HC, Katsarava Z. Symptomatic Migraine and Pontine Vascular Malformation. Cephalalgia 2016; 26:763-6. [PMID: 16686921 DOI: 10.1111/j.1468-2982.2006.01102.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- M Obermann
- Department of Neurology, University Duisburg-Essen, Essen, Germany.
| | | | | | | | | |
Collapse
|
37
|
Askenase MH, Sansing LH. Stages of the Inflammatory Response in Pathology and Tissue Repair after Intracerebral Hemorrhage. Semin Neurol 2016; 36:288-97. [PMID: 27214704 DOI: 10.1055/s-0036-1582132] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major health concern, with high rates of mortality and morbidity and no highly effective clinical interventions. Basic research in animal models of ICH has provided insight into its complex pathology, in particular revealing the role of inflammation in driving neuronal death and neurologic deficits after hemorrhage. The response to ICH occurs in four distinct phases: (1) initial tissue damage and local activation of inflammatory factors, (2) inflammation-driven breakdown of the blood-brain barrier, (3) recruitment of circulating inflammatory cells and subsequent secondary immunopathology, and (4) engagement of tissue repair responses that promote tissue repair and restoration of neurologic function. The development of CNS inflammation occurs over many days after initial hemorrhage and thus may represent an ideal target for treatment of the disease, but further research is required to identify the mechanisms that promote engagement of inflammatory versus anti-inflammatory pathways. In this review, the authors examine how experimental models of ICH have uncovered critical mediators of pathology in each of the four stages of the inflammatory response, and focus on the role of the immune system in these processes.
Collapse
Affiliation(s)
- Michael H Askenase
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
38
|
Galho AR, Cordeiro MF, Ribeiro SA, Marques MS, Antunes MFD, Luz DC, Hädrich G, Muccillo-Baisch AL, Barros DM, Lima JV, Dora CL, Horn AP. Protective role of free and quercetin-loaded nanoemulsion against damage induced by intracerebral haemorrhage in rats. NANOTECHNOLOGY 2016; 27:175101. [PMID: 26965041 DOI: 10.1088/0957-4484/27/17/175101] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Intracerebral haemorrhage (ICH) is a worldwide public health problem. Experimental studies have shown that oxidative stress plays an important role in the pathogenesis of ICH and could represent a target for its treatment. However, the blood-brain barrier is an obstacle to be overcome, as it hampers the administration of compounds to the central nervous system. In this study, we compared the effects of a quercetin-loaded nanoemulsion (QU-N) with the free form of the drug (QU-SP) in a collagenase-induced ICH rat model. Quercetin (QU) is a polyphenol that has an antioxidant effect in vitro, but due to its high lipophilicity, it has low bioavailability in vivo. In this study, animals submitted or not to ICH were treated with a single intraperitoneal QU dose (free or nanoemulsion) of 30 mg kg(-1). Motor assessment was evaluated by the open field, foot fault and beam walking behavioural tests. 72 h after surgery the haematoma size was evaluated and biochemical measurements were performed. Animals treated with QU-N had a significant improvement in the beam walking and open field tests. Also, QU-N was able to reduce the size of the haematoma, preserving the activity of glutathione S-transferase (GST), increasing GSH content, and the total antioxidant capacity. QU-SP recovered locomotor activity and increased the GSH content and the total antioxidant capacity. Thus, it can be observed that QU presented antioxidant activity in both formulations, but the incorporation into nanoemulsions increased its antioxidant effect, which was reflected in the improvement of the motor skills and in the haematoma size decrement. These results suggest that the nanoemulsion containing QU developed in this study could be promising for future studies on treatments for ICH.
Collapse
Affiliation(s)
- A R Galho
- Programa de Pós-graduação em Ciências Fisiológicas - Fisiologia Animal Comparada, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, 96210-900, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Intracerebral Hemorrhage, Oxidative Stress, and Antioxidant Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1203285. [PMID: 27190572 PMCID: PMC4848452 DOI: 10.1155/2016/1203285] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/20/2015] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
Hemorrhagic stroke is a common and severe neurological disorder and is associated with high rates of mortality and morbidity, especially for intracerebral hemorrhage (ICH). Increasing evidence demonstrates that oxidative stress responses participate in the pathophysiological processes of secondary brain injury (SBI) following ICH. The mechanisms involved in interoperable systems include endoplasmic reticulum (ER) stress, neuronal apoptosis and necrosis, inflammation, and autophagy. In this review, we summarized some promising advances in the field of oxidative stress and ICH, including contained animal and human investigations. We also discussed the role of oxidative stress, systemic oxidative stress responses, and some research of potential therapeutic options aimed at reducing oxidative stress to protect the neuronal function after ICH, focusing on the challenges of translation between preclinical and clinical studies, and potential post-ICH antioxidative therapeutic approaches.
Collapse
|
40
|
Microglial Polarization and Inflammatory Mediators After Intracerebral Hemorrhage. Mol Neurobiol 2016; 54:1874-1886. [PMID: 26894396 DOI: 10.1007/s12035-016-9785-6] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 02/08/2016] [Indexed: 12/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke with high mortality and morbidity. When a diseased artery within the brain bursts, expansion and absorption of the resulting hematoma trigger a series of reactions that cause primary and secondary brain injury. Microglia are extremely important for removing the hematoma and clearing debris, but they are also a source of ongoing inflammation. This article discusses the role of microglial activation/polarization and related inflammatory mediators, such as Toll-like receptor 4, matrix metalloproteinases, high-mobility group protein box-1, nuclear factor erythroid 2-related factor 2, heme oxygenase, and iron, in secondary injury after ICH and highlights the potential targets for ICH treatment.
Collapse
|
41
|
Yu QJ, Tao H, Wang X, Li MC. Targeting brain microvascular endothelial cells: a therapeutic approach to neuroprotection against stroke. Neural Regen Res 2016; 10:1882-91. [PMID: 26807131 PMCID: PMC4705808 DOI: 10.4103/1673-5374.170324] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Brain microvascular endothelial cells form the interface between nervous tissue and circulating blood, and regulate central nervous system homeostasis. Brain microvascular endothelial cells differ from peripheral endothelial cells with regards expression of specific ion transporters and receptors, and contain fewer fenestrations and pinocytotic vesicles. Brain microvascular endothelial cells also synthesize several factors that influence blood vessel function. This review describes the morphological characteristics and functions of brain microvascular endothelial cells, and summarizes current knowledge regarding changes in brain microvascular endothelial cells during stroke progression and therapies. Future studies should focus on identifying mechanisms underlying such changes and developing possible neuroprotective therapeutic interventions.
Collapse
Affiliation(s)
- Qi-Jin Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Hong Tao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ming-Chang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
42
|
Association Between High Arterial Oxygen Tension and Long-Term Survival After Spontaneous Intracerebral Hemorrhage. Crit Care Med 2016; 44:180-7. [DOI: 10.1097/ccm.0000000000001281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Mechanisms of Cerebral Hemorrhage. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
Oxidative Stress in Intracerebral Hemorrhage: Sources, Mechanisms, and Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3215391. [PMID: 26843907 PMCID: PMC4710930 DOI: 10.1155/2016/3215391] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is associated with the highest mortality and morbidity despite only constituting approximately 10–15% of all strokes. Complex underlying mechanisms consisting of cytotoxic, excitotoxic, and inflammatory effects of intraparenchymal blood are responsible for its highly damaging effects. Oxidative stress (OS) also plays an important role in brain injury after ICH but attracts less attention than other factors. Increasing evidence has demonstrated that the metabolite axis of hemoglobin-heme-iron is the key contributor to oxidative brain damage after ICH, although other factors, such as neuroinflammation and prooxidases, are involved. This review will discuss the sources, possible molecular mechanisms, and potential therapeutic targets of OS in ICH.
Collapse
|
45
|
Tang J, Tao Y, Jiang B, Chen Q, Hua F, Zhang J, Zhu G, Chen Z. Pharmacological Preventions of Brain Injury Following Experimental Germinal Matrix Hemorrhage: an Up-to-Date Review. Transl Stroke Res 2015; 7:20-32. [PMID: 26561051 DOI: 10.1007/s12975-015-0432-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 10/24/2015] [Accepted: 11/02/2015] [Indexed: 12/19/2022]
Abstract
Germinal matrix hemorrhage (GMH) is defined as the rupture of immature blood vessels in the subependymal zone of premature infants with significant mortality and morbidity. Considering the notable social and ecological stress brought by GMH-induced brain injury and sequelae, safe and efficient pharmacological preventions are badly needed. Currently, several appropriate animal models are available to mimic the clinical outcomes of GMH in human patients. In the long run, hemorrhagic strokes are the research target. Previously, we found that minocycline was efficient to alleviate GMH-induced brain edema and posthemorrhagic hydrocephalus (PHH) in rats, which may be closely related to the activation of cannabinoid receptor 2 (CB2R). However, how the two molecules correlate and the underlined molecular pathway remain unknown. To extensively understand current experimental GMH treatment, this literature review critically evaluates existing therapeutic strategies, potential treatments, and potentially involved molecular mechanisms. Each strategy has its own advantages and disadvantages. Some of the mechanisms are still controversial, requiring an increasing number of animal experiments before the therapeutic strategy would be widely accepted.
Collapse
Affiliation(s)
- Jun Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Yihao Tao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Bing Jiang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Feng Hua
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - John Zhang
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China.
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
46
|
Xiong XY, Chen J, Zhu WY, Zhao T, Zhong Q, Zhou K, Meng ZY, Wang YC, Wang PF, Fang H, Yang QW. Serum hepcidin concentrations correlate with serum iron level and outcome in patients with intracerebral hemorrhage. Neurol Sci 2015; 36:1843-9. [PMID: 26024728 DOI: 10.1007/s10072-015-2266-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/22/2015] [Indexed: 12/20/2022]
Abstract
Iron plays a detrimental role in the intracerebral hemorrhage (ICH)-induced brain damage, while hepcidin is the most important iron-regulated hormone. Here, we investigate the association between serum hepcidin and serum iron, outcome in patients with ICH. Serum samples of 81 cases with ICH were obtained on consecutive days to detect the levels of hepcidin, iron, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). The National Institutes of Health Stroke Scale score (NIHSS) was measured at admission and on days 7 and 30, and the modified Rankin Scale (mRS) score was evaluated at 3 months after ICH. Additionally, the correlations of serum hepcidin with serum iron and the mRS score were analyzed by a generalized linear model. Higher serum hepcidin levels were detected in patients with poor outcomes (P < 0.001), and the mRS score increased by a mean of 1.135 points (95% CI 1.021-1.247, P < 0.001) for every serum hepcidin quartile after adjusting for other prognostic variables. Pearson correlation analysis showed that serum hepcidin was negatively correlated with serum iron (r = -0.5301, P < 0.001), and a significantly lower concentration of serum iron was found in patients with poor outcomes (P = 0.007). Additionally, serum hepcidin was independently correlated with mRS scores of ICH patients (OR 1.115, 95% CI 0.995-1.249, P = 0.021). Our results suggest that serum hepcidin is closely related to the outcome of patients with ICH and may be a biological marker for outcome prediction.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Jing Chen
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Wen-Yao Zhu
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Ting Zhao
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Qi Zhong
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Kai Zhou
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Zhao-You Meng
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Yan-Chun Wang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Peng-Fei Wang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Huang Fang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, No. 183, Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
47
|
Egashira Y, Hua Y, Keep RF, Xi G. Intercellular cross-talk in intracerebral hemorrhage. Brain Res 2015; 1623:97-109. [PMID: 25863131 DOI: 10.1016/j.brainres.2015.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disorder with high mortality and morbidity. Currently, there are few treatment strategies for ICH-induced brain injury. A recent increase in interest in the pathophysiology of ICH has led to elucidation of the pathways underlying ICH-induced brain injury, pathways where intercellular and hematoma to cell signaling play important roles. In this review, we summarize recent advances in ICH research focusing on intercellular and hematoma:cell cross-talk related to brain injury and recovery after ICH. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yusuke Egashira
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
48
|
Zhao XR, Gonzales N, Aronowski J. Pleiotropic role of PPARγ in intracerebral hemorrhage: an intricate system involving Nrf2, RXR, and NF-κB. CNS Neurosci Ther 2014; 21:357-66. [PMID: 25430543 DOI: 10.1111/cns.12350] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/10/2014] [Accepted: 10/11/2014] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke involving formation of hematoma within brain parenchyma, which accounts for 8-15% of all strokes in Western societies and 20-30% among Asian populations, and has a 1-year mortality rate >50%. The high mortality and severe morbidity make ICH a major public health problem. Only a few evidence-based targeted treatments are used for ICH management, and interventions focus primarily on supportive care and comorbidity prevention. Even in patients who survive the ictus, extravasated blood (including plasma components) and subsequent intrahematoma hemolytic products trigger a series of adverse events within the brain parenchyma, leading to secondary brain injury, edema and severe neurological deficits or death. Although the hematoma in humans gradually resolves within months, full restoration of neurological function can be slow and often incomplete, leaving survivors with devastating neurological deficits. During past years, peroxisome proliferator-activated receptor gamma (PPARγ) transcription factor and its agonists received recognition as important players in regulating not only glucose and lipid metabolism (which underlies its therapeutic effect in type 2 diabetes mellitus), and more recently, as an instrumental pleiotropic regulator of antiinflammation, antioxidative regulation, and phagocyte-mediated cleanup processes. PPARγ agonists have emerged as potential therapeutic target for stroke. The use of PPARγ as a therapeutic target appears to have particularly strong compatibility toward pathogenic components of ICH. In addition to its direct genomic effect, PPARγ may interact with transcription factor, NF-κB, which may underlie many aspects of the antiinflammatory effect of PPARγ. Furthermore, PPARγ appears to regulate expression of Nrf2, another transcription factor and master regulator of detoxification and antioxidative regulation. Finally, the synergistic costimulation of PPARγ and retinoid X receptor, RXR, may play an additional role in the therapeutic modulation of PPARγ function. In this article, we outline the main components of the role of PPARγ in ICH pathogenesis.
Collapse
Affiliation(s)
- Xiu-Rong Zhao
- Department of Neurology, Stroke Research Center, University of Texas Medical School - Houston, Houston, TX, USA
| | | | | |
Collapse
|
49
|
Shang H, Cui D, Yang D, Liang S, Zhang W, Zhao W. The radical scavenger edaravone improves neurologic function and perihematomal glucose metabolism after acute intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2014; 24:215-22. [PMID: 25440340 DOI: 10.1016/j.jstrokecerebrovasdis.2014.08.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 07/29/2014] [Accepted: 08/22/2014] [Indexed: 12/30/2022] Open
Abstract
Oxidative injury caused by reactive oxygen species plays an important role in the progression of intracerebral hemorrhage (ICH)-induced secondary brain injury. Previous studies have demonstrated that the free radical scavenger edaravone may prevent neuronal injury and brain edema after ICH. However, the influence of edaravone on cerebral metabolism in the early stages after ICH and the underlying mechanism have not been fully investigated. In the present study, we investigated the effect of edaravone on perihematomal glucose metabolism using (18)F-fluorordeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT). Additionally, the neurologic deficits, brain edemas, and cell death that followed ICH were quantitatively analyzed. After blood infusion, the rats treated with edaravone showed significant improvement in both forelimb placing and corner turn tests compared with those treated with vehicle. Moreover, the brain water content of the edaravone-treated group was significantly decreased compared with that of the vehicle group on day 3 after ICH. PET/CT images of ICH rats exhibited obvious decreases in FDG standardized uptake values in perihematomal region on day 3, and the lesion-to-normal ratio of the edaravone-treated ICH rats was significantly increased compared with that of the control rats. Calculation of the brain injury volumes from the PET/CT images revealed that the volumes of the blood-induced injuries were significantly smaller in the edaravone group compared with the vehicle group. Terminal Deoxynucleotidyl Transferase-mediated dUTP Nick End Labeling assays performed 3 days after ICH revealed that the numbers of apoptotic cells in perihematomal region of edaravone-treated ICH rats were decreased relative to the vehicle group. Thus, the present study demonstrates that edaravone has scavenging properties that attenuate neurologic behavioral deficits and brain edema in the early period of ICH. Additionally, edaravone may improve cerebral metabolism around the hematoma by attenuating apoptotic cell death after ICH.
Collapse
Affiliation(s)
- Hanbing Shang
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Derong Cui
- Department of Anesthesiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dehua Yang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng Liang
- Department of Nuclear Medicine and Micro PET/CT Research Center, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weifeng Zhang
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiguo Zhao
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
50
|
Das S, Roy S, Kaul S, Jyothy A, Munshi A. E-selectin gene (S128R) polymorphism in hemorrhagic stroke: Comparison with ischemic stroke. Neurosci Lett 2014; 581:125-8. [DOI: 10.1016/j.neulet.2014.08.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 07/24/2014] [Accepted: 08/18/2014] [Indexed: 11/17/2022]
|